Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 592
Filtrar
1.
Arch Dermatol Res ; 315(7): 2003-2009, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36877308

RESUMO

BACKGROUND: Lymphocyte enhancer-binding factor-1 (LEF1) is responsible for melanocyte proliferation, migration and differentiation and its downregulation may result in depigmentation in vitiligo. Narrowband UVB (NB-UVB) phototherapy is known to enhance melanocyte migration from hair follicles to lesional epidermis; hence, it may have a role in the upregulation of LEF1. OBJECTIVES: We intended to assess the expression of LEF1 both before and after NB-UVB therapy and correlate it with the extent of re-pigmentation. MATERIALS AND METHODS: In this prospective cohort study, 30 patients of unstable non-segmental vitiligo were administered NB-UVB phototherapy for 24 weeks. Skin biopsies were obtained from acral and non-acral sites in all patients, both prior to initiation and after completion of phototherapy and LEF1 expression was measured. RESULTS: Amongst the 16 patients who completed the study, at 24 weeks, all patients achieved > 50% re-pigmentation. However, > 75% re-pigmentation was achieved in only 11.1% of acral patches, whereas it was achieved in a significantly higher number of non-acral patches (66.6%) (p = 0.05). A significant increase was observed in the mean fluorescent intensity of the LEF1 gene in both acral as well as non-acral areas at 24 weeks as compared to baseline (p = 0.0078), However, no difference was observed between acral and non-acral lesions in the LEF1 expression at 24 weeks or the change in LEF1 expression from baseline. CONCLUSION: LEF1 expression modulates the re-pigmentation of vitiligo lesions after treatment with NBUVB phototherapy.


Assuntos
Fator 1 de Ligação ao Facilitador Linfoide , Pigmentação , Vitiligo , Fator 1 de Ligação ao Facilitador Linfoide/genética , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Raios Ultravioleta , Fototerapia/efeitos adversos , Fototerapia/normas , Vitiligo/genética , Vitiligo/terapia , Humanos , Masculino , Feminino , Adulto , Pessoa de Meia-Idade , Pigmentação/genética , Pigmentação/efeitos da radiação , Regulação para Cima/efeitos da radiação , Estudos Prospectivos , Índia , Imuno-Histoquímica
2.
J Photochem Photobiol B ; 241: 112670, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36841175

RESUMO

BACKGROUND: Antimicrobial blue light (aBL) kills a variety of bacteria, including Porphyromonas gingivalis. However, little is known about the transcriptomic response of P. gingivalis to aBL therapy. This study was designed to evaluate the selective cytotoxicity of aBL against P. gingivalis over human cells and to further investigate the genetic response of P. gingivalis to aBL at the transcriptome level. METHODS: Colony forming unit (CFU) testing, confocal laser scanning microscopy (CLSM), and scanning electron microscopy (SEM) were used to investigate the antimicrobial effectiveness of blue light against P. gingivalis. The temperatures of the irradiated targets were measured to prevent overheating. Multiple fluorescent probes were used to quantify reactive oxygen species (ROS) generation after blue-light irradiation. RNA sequencing (RNA-seq) was used to investigate the changes in global gene expression. Following the screening of target genes, real-time quantitative polymerase chain reaction (RT-qPCR) was performed to confirm the regulation of gene expression. RESULTS: A 405 nm aBL at 100 mW/cm2 significantly killed P. gingivalis within 5 min while sparing human gingival fibroblasts (HGFs). No obvious temperature changes were detected in the irradiated surface under our experimental conditions. RNA-seq showed that the transcription of multiple genes was regulated, and RT-qPCR revealed that the expression levels of the genes RgpA and RgpB, which may promote heme uptake, as well as the genes Ftn and FetB, which are related to iron homeostasis, were significantly upregulated. The expression levels of the FeoB-2 and HmuR genes, which are related to hydroxyl radical scavenging, were significantly downregulated. CONCLUSIONS: aBL strengthens the heme uptake and iron export gene pathways while reducing the ROS scavenging pathways in P. gingivalis, thus improving the accumulation of endogenous photosensitizers and enhancing oxidative damage to P. gingivalis.


Assuntos
Cor , Regulação Bacteriana da Expressão Gênica , Genes Bacterianos , Ferro , Luz , Porfirinas , Porphyromonas gingivalis , Porfirinas/metabolismo , Ferro/metabolismo , Porphyromonas gingivalis/citologia , Porphyromonas gingivalis/genética , Porphyromonas gingivalis/metabolismo , Porphyromonas gingivalis/efeitos da radiação , Transporte Biológico/genética , Transporte Biológico/efeitos da radiação , Humanos , Gengiva/citologia , Fibroblastos/citologia , Fibroblastos/efeitos da radiação , Radical Hidroxila/metabolismo , Heme/metabolismo , Regulação para Cima/efeitos da radiação , Homeostase/efeitos da radiação , Regulação para Baixo/efeitos da radiação , Viabilidade Microbiana/efeitos da radiação , Espécies Reativas de Oxigênio/metabolismo , Aerobiose , Genes Bacterianos/efeitos da radiação , Regulação Bacteriana da Expressão Gênica/genética , Regulação Bacteriana da Expressão Gênica/efeitos da radiação
3.
Int J Mol Sci ; 23(2)2022 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-35055167

RESUMO

The receptor tyrosine kinase AXL (RTK-AXL) is implicated in therapy resistance and tumor progression in glioblastoma multiforme (GBM). Here, we investigated therapy-induced receptor modifications and how endogenous RTK-AXL expression and RTK-AXL inhibition contribute to therapy resistance in GBM. GBM cell lines U118MG and SF126 were exposed to temozolomide (TMZ) and radiation (RTX). Receptor modifications in response to therapy were investigated on protein and mRNA levels. TMZ-resistant and RTK-AXL overexpressing cell lines were exposed to increasing doses of TMZ and RTX, with and without RTK-AXL tyrosine kinase inhibitor (TKI). Colorimetric microtiter (MTT) assay and colony formation assay (CFA) were used to assess cell viability. Results showed that the RTK-AXL shedding product, C-terminal AXL (CT-AXL), rises in response to repeated TMZ doses and under hypoxia, acts as a surrogate marker for radio-resistance. Endogenous RTX-AXL overexpression leads to therapy resistance, whereas combination therapy of TZM and RTX with TKI R428 significantly increases therapeutic effects. This data proves the role of RTK-AXL in acquired and intrinsic therapy resistance. By demonstrating that therapy resistance may be overcome by combining AXL TKI with standard treatments, we have provided a rationale for future study designs investigating AXL TKIs in GBM.


Assuntos
Benzocicloeptenos/farmacologia , Neoplasias Encefálicas/genética , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Glioblastoma/genética , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , Temozolomida/farmacologia , Triazóis/farmacologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/terapia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Terapia Combinada , Resistencia a Medicamentos Antineoplásicos/efeitos da radiação , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Glioblastoma/metabolismo , Glioblastoma/terapia , Humanos , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/efeitos da radiação , Hipóxia Tumoral/efeitos dos fármacos , Hipóxia Tumoral/efeitos da radiação , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/efeitos da radiação , Receptor Tirosina Quinase Axl
4.
PLoS Biol ; 19(11): e3001471, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34788294

RESUMO

Trichoplax adhaerens is the simplest multicellular animal with tissue differentiation and somatic cell turnover. Like all other multicellular organisms, it should be vulnerable to cancer, yet there have been no reports of cancer in T. adhaerens or any other placozoan. We investigated the cancer resistance of T. adhaerens, discovering that they are able to tolerate high levels of radiation damage (218.6 Gy). To investigate how T. adhaerens survive levels of radiation that are lethal to other animals, we examined gene expression after the X-ray exposure, finding overexpression of genes involved in DNA repair and apoptosis including the MDM2 gene. We also discovered that T. adhaerens extrudes clusters of inviable cells after X-ray exposure. T. adhaerens is a valuable model organism for studying the molecular, genetic, and tissue-level mechanisms underlying cancer suppression.


Assuntos
Reparo do DNA/genética , Placozoa/genética , Tolerância a Radiação/genética , Regulação para Cima/genética , Animais , Dano ao DNA/genética , Dano ao DNA/efeitos da radiação , Reparo do DNA/efeitos da radiação , Regulação da Expressão Gênica/efeitos da radiação , Placozoa/anatomia & histologia , Placozoa/efeitos da radiação , Exposição à Radiação , Análise de Sequência de DNA , Regulação para Cima/efeitos da radiação , Sequenciamento Completo do Genoma , Raios X
5.
Cells ; 10(8)2021 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-34440856

RESUMO

The primary cause of colorectal cancer (CRC) recurrence is increased distant metastasis after radiotherapy, so there is a need for targeted therapeutic approaches to reduce the metastatic-relapse risk. Dysregulation of the cell-surface glycoprotein podocalyxin-like protein (PODXL) plays an important role in promoting cancer-cell motility and is associated with poor prognoses for many malignancy types. We found that CRC cells exposed to radiation demonstrated increased TGFß and PODXL expressions, resulting in increased migration and invasiveness due to increased extracellular matrix deposition. In addition, both TGFß and PODXL were highly expressed in tissue samples from radiotherapy-treated CRC patients compared to those from patients without this treatment. However, it is unclear whether TGFß and PODXL interactions are involved in cancer-progression resistance after radiation exposure in CRC. Here, using CRC cells, we showed that silencing PODXL blocked radiation-induced cell migration and invasiveness. Cell treatment with galunisertib (a TGFß-pathway inhibitor) also led to reduced viability and migration, suggesting that its clinical use may enhance the cytotoxic effects of radiation and lead to the effective inhibition of CRC progression. Overall, the results demonstrate that downregulation of TGFß and its-mediated PODXL may provide potential therapeutic targets for patients with radiotherapy-resistant CRC.


Assuntos
Neoplasias Colorretais/patologia , Radiação Ionizante , Sialoglicoproteínas/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Regulação para Cima/efeitos da radiação , Caderinas/genética , Caderinas/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/efeitos da radiação , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Neoplasias Colorretais/metabolismo , Transição Epitelial-Mesenquimal/genética , Transição Epitelial-Mesenquimal/efeitos da radiação , Humanos , Metástase Neoplásica , Prognóstico , Pirazóis/farmacologia , Quinolinas/farmacologia , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Sialoglicoproteínas/antagonistas & inibidores , Sialoglicoproteínas/genética , Fator de Crescimento Transformador beta/antagonistas & inibidores , Vimentina/genética , Vimentina/metabolismo
6.
Int J Mol Sci ; 22(16)2021 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-34445085

RESUMO

Retinal ganglion cells (RGCs) undergo dendritic pruning in a variety of neurodegenerative diseases, including glaucoma and autosomal dominant optic atrophy (ADOA). Axotomising RGCs by severing the optic nerve generates an acute model of RGC dendropathy, which can be utilized to assess the therapeutic potential of treatments for RGC degeneration. Photobiomodulation (PBM) with red light provided neuroprotection to RGCs when administered ex vivo to wild-type retinal explants. In the current study, we used aged (13-15-month-old) wild-type and heterozygous B6;C3-Opa1Q285STOP (Opa1+/-) mice, a model of ADOA exhibiting RGC dendropathy. These mice were pre-treated with 4 J/cm2 of 670 nm light for five consecutive days before the eyes were enucleated and the retinas flat-mounted into explant cultures for 0-, 8- or 16-h ex vivo. RGCs were imaged by confocal microscopy, and their dendritic architecture was quantified by Sholl analysis. In vivo 670 nm light pretreatment inhibited the RGC dendropathy observed in untreated wild-type retinas over 16 h ex vivo and inhibited dendropathy in ON-center RGCs in wild-type but not Opa1+/- retinas. Immunohistochemistry revealed that aged Opa1+/- RGCs exhibited increased nitrosative damage alongside significantly lower activation of NF-κB and upregulation of DJ-1. PBM restored NF-κB activation in Opa1+/- RGCs and enhanced DJ-1 expression in both genotypes, indicating a potential molecular mechanism priming the retina to resist future oxidative insult. These data support the potential of PBM as a treatment for diseases involving RGC degeneration.


Assuntos
Atrofia Óptica Autossômica Dominante/terapia , Fototerapia , Proteína Desglicase DJ-1/análise , Células Ganglionares da Retina/patologia , Células Ganglionares da Retina/efeitos da radiação , Animais , Modelos Animais de Doenças , Luz , Camundongos , Neuroproteção/efeitos da radiação , Atrofia Óptica Autossômica Dominante/patologia , Degeneração Retiniana , Células Ganglionares da Retina/citologia , Regulação para Cima/efeitos da radiação
7.
J Radiat Res ; 62(5): 773-781, 2021 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-34196706

RESUMO

Programmed death ligand 1 (PD-L1) expression on the surface of cancer cells affects the efficacy of anti-PD-1/PD-L1 immune checkpoint therapy. However, the mechanism underlying PD-L1 expression in cancer cells is not fully understood, particularly after ionizing radiation (IR). Here, we examined the impact of high linear energy transfer (LET) carbon-ion irradiation on the expression of PD-L1 in human osteosarcoma U2OS cells. We found that the upregulation of PD-L1 expression after high LET carbon-ion irradiation was greater than that induced by X-rays at the same physical and relative biological effectiveness (RBE) dose, and that the upregulation of PD-L1 induced by high LET carbon-ion irradiation was predominantly dependent on ataxia telangiectasia and Rad3-related (ATR) kinase activity. Moreover, we showed that the downstream signaling, e.g. STAT1 phosphorylation and IRF1 expression, was upregulated to a greater extent after high LET carbon-ion irradiation than X-rays, and that IRF1 upregulation was also ATR dependent. Finally, to visualize PD-L1 molecules on the cell surface in 3D, we applied immunofluorescence-based super-resolution imaging. The three-dimensional structured illumination microscopy (3D-SIM) analyses revealed substantial increases in the number of presented PD-L1 molecules on the cell surface after high LET carbon-ion irradiation compared with X-ray irradiation.


Assuntos
Antígeno B7-H1/biossíntese , Neoplasias Ósseas/patologia , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Radioterapia com Íons Pesados , Proteínas de Neoplasias/biossíntese , Osteossarcoma/patologia , Proteínas Mutadas de Ataxia Telangiectasia/antagonistas & inibidores , Proteínas Mutadas de Ataxia Telangiectasia/fisiologia , Antígeno B7-H1/genética , Linhagem Celular Tumoral , Humanos , Imageamento Tridimensional , Fator Regulador 1 de Interferon/biossíntese , Fator Regulador 1 de Interferon/genética , Transferência Linear de Energia , Morfolinas/farmacologia , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Fosforilação/efeitos da radiação , Processamento de Proteína Pós-Traducional/efeitos da radiação , Pirazinas/farmacologia , Pironas/farmacologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Neoplásico/biossíntese , RNA Neoplásico/genética , Fator de Transcrição STAT1/metabolismo , Sulfonas/farmacologia , Regulação para Cima/efeitos da radiação , Raios X
8.
Cancer Sci ; 112(10): 4064-4074, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34251713

RESUMO

Programmed cell death ligand 1 (PD-L1) is a major immunosuppressive checkpoint protein expressed by tumor cells to subvert anticancer immunity. Recent studies have shown that ionizing radiation (IR) upregulates the expression of PD-L1 in tumor cells. However, whether an IR-induced DNA damage response (DDR) directly regulates PD-L1 expression and the functional significance of its upregulation are not fully understood. Here, we show that IR-induced upregulation of PD-L1 expression proceeds through both transcriptional and post-translational mechanisms. Upregulated PD-L1 was predominantly present on the cell membrane, resulting in T-cell apoptosis in a co-culture system. Using mass spectrometry, we identified PD-L1 interacting proteins and found that BCLAF1 (Bcl2 associated transcription factor 1) is an important regulator of PD-L1 in response to IR. BCLAF1 depletion decreased PD-L1 expression by promoting the ubiquitination of PD-L1. In addition, we show that CMTM6 is upregulated in response to IR and participates in BCLAF1-dependent PD-L1 upregulation. Finally, we demonstrated that the ATM/BCLAF1/PD-L1 axis regulated PD-L1 stabilization in response to IR. Together, our findings reveal a novel regulatory mechanism of PD-L1 expression in the DDR.


Assuntos
Antígeno B7-H1/metabolismo , Radiação Ionizante , Proteínas Repressoras/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Apoptose , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Antígeno B7-H1/efeitos da radiação , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Técnicas de Cocultura , Dano ao DNA , Humanos , Células Jurkat , Proteínas com Domínio MARVEL/metabolismo , Proteínas com Domínio MARVEL/efeitos da radiação , Espectrometria de Massas , Proteínas da Mielina/metabolismo , Proteínas da Mielina/efeitos da radiação , Proteínas de Neoplasias/metabolismo , Modificação Traducional de Proteínas , Processamento de Proteína Pós-Traducional , Proteínas Repressoras/deficiência , Linfócitos T/citologia , Linfócitos T/efeitos da radiação , Proteínas Supressoras de Tumor/deficiência , Ubiquitinação , Regulação para Cima/efeitos da radiação
9.
J Photochem Photobiol B ; 221: 112243, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34217028

RESUMO

Due to their capacity to differentiate into the chondrogenic lineage, adipose-derived stromal/stem cells (ASC) are a promising source of therapeutically relevant cells for cartilage tissue regeneration. Their differentiation potential, however, varies between patients. In our study, we aim to stimulate ASC towards a more reliable chondrogenic phenotype using photobiomodulation (PBM). LED devices of either blue (475 nm), green (516 nm) or red (635 nm) light were used to treat human ASC from donors of varying chondrogenic potential. The treatment was applied either once during the 2D expansion phase or repeatedly during the 3D differentiation phase. Chondrogenic differentiation was assessed via pellet size, GAG/DNA content, histology and gene expression analysis. Reactions to PBM were found to be wavelength-dependent and more pronounced when the treatment was applied during expansion. Donors were assigned to responder categories according to their response to the treatment during expansion, whereby good responders were mainly donors with low intrinsic chondrogenic potential. Exposed to light, they revealed a particularly high relative increase in pellet size (more than twice the size of untreated controls after red light PBM), intense collagen type II immunostaining (low/absent in untreated controls) and activation of otherwise absent COL2A1 expression. Conversely, on a donor with high intrinsic chondrogenic potential, light had adverse effects. When applied with shorter wavelengths (blue, green), it led to reduced pellet size, GAG/DNA content and collagen type II immunostaining. However, when PBM was applied in 3D, the same donor was the only one to react with increased differentiation to all three wavelengths. We were able to demonstrate that PBM can be used to enhance or hamper chondrogenesis of ASC, and that success depends on treatment parameters and intrinsic cellular potential. The improvement of chondrogenesis in donors with low intrinsic potential highlights PBM as potent tool for cell-based cartilage regeneration. Its cost-effectiveness and ease of use make for an attractive treatment option to enhance the performance of ASC in cartilage tissue engineering.


Assuntos
Diferenciação Celular/efeitos da radiação , Condrogênese/efeitos da radiação , Luz , Tecido Adiposo/citologia , Técnicas de Cultura de Células , Células Cultivadas , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Regulação para Baixo/efeitos da radiação , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Regulação para Cima/efeitos da radiação
10.
Cancer Cell ; 39(9): 1202-1213.e6, 2021 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-34329585

RESUMO

Studies suggest that the efficacy of cancer chemotherapy and immunotherapy is influenced by intestinal bacteria. However, the influence of the microbiome on radiation therapy is not as well understood, and the microbiome comprises more than bacteria. Here, we find that intestinal fungi regulate antitumor immune responses following radiation in mouse models of breast cancer and melanoma and that fungi and bacteria have opposite influences on these responses. Antibiotic-mediated depletion or gnotobiotic exclusion of fungi enhances responsiveness to radiation, whereas antibiotic-mediated depletion of bacteria reduces responsiveness and is associated with overgrowth of commensal fungi. Further, elevated intratumoral expression of Dectin-1, a primary innate sensor of fungi, is negatively associated with survival in patients with breast cancer and is required for the effects of commensal fungi in mouse models of radiation therapy.


Assuntos
Antifúngicos/administração & dosagem , Bactérias/classificação , Neoplasias da Mama/terapia , Fungos/efeitos dos fármacos , Lectinas Tipo C/genética , Melanoma/terapia , Animais , Antifúngicos/farmacologia , Bactérias/imunologia , Neoplasias da Mama/imunologia , Neoplasias da Mama/microbiologia , Terapia Combinada , Regulação para Baixo , Feminino , Fungos/classificação , Fungos/imunologia , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/efeitos da radiação , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos , Melanoma/imunologia , Melanoma/microbiologia , Camundongos , Simbiose , Linfócitos T/metabolismo , Macrófagos Associados a Tumor/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/efeitos da radiação , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Int J Mol Sci ; 22(14)2021 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-34298925

RESUMO

The Linear-No-Threshold (LNT) model predicts a dose-dependent linear increase in cancer risk. This has been supported by biological and epidemiological studies at high-dose exposures. However, at low-doses (LDR ≤ 0.1 Gy), the effects are more elusive and demonstrate a deviation from linearity. In this study, the effects of LDR on the development and progression of mammary cancer in FVB/N-Tg(MMTVneu)202Mul/J mice were investigated. Animals were chronically exposed to total doses of 10, 100, and 2000 mGy via tritiated drinking water, and were assessed at 3.5, 6, and 8 months of age. Results indicated an increased proportion of NK cells in various organs of LDR exposed mice. LDR significantly influenced NK and T cell function and activation, despite diminishing cell proliferation. Notably, the expression of NKG2D receptor on NK cells was dramatically reduced at 3.5 months but was upregulated at later time-points, while the expression of NKG2D ligand followed the opposite trend, with an increase at 3.5 months and a decrease thereafter. No noticeable impact was observed on mammary cancer development, as measured by tumor load. Our results demonstrated that LDR significantly influenced the proportion, proliferation, activation, and function of immune cells. Importantly, to the best of our knowledge, this is the first report demonstrating that LDR modulates the cross-talk between the NKG2D receptor and its ligands.


Assuntos
Neoplasias da Mama/imunologia , Neoplasias da Mama/radioterapia , Imunidade/efeitos da radiação , Animais , Neoplasias da Mama/metabolismo , Proliferação de Células/efeitos da radiação , Feminino , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/efeitos da radiação , Ligantes , Camundongos , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Doses de Radiação , Linfócitos T/metabolismo , Linfócitos T/efeitos da radiação , Regulação para Cima/efeitos da radiação
12.
Int J Radiat Biol ; 97(9): 1324-1332, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34125651

RESUMO

PURPOSE: Recently, therapeutic effects of extremely low-frequency electromagnetic field (ELF-EMF) as complementary and alternative medicine, used in the oncology field to control disease symptoms. Micro RNAs (miRs) are responsible for the post-transcriptional regulation of gene expression in the cell. This study aimed to evaluate the expression changes of miR-144 and miR-375 in the human gastric adenocarcinoma cell line (AGS) under the exposure of ELF-EMF. MATERIALS AND METHODS: AGS cells were exposed to magnetic flux densities of 0.2 and 2 mT for 18 h, continuously and discontinuously (1.5 h on/1.5 h off). Cell viability was evaluated by MTT assay. Changes of miR-144 expression levels in AGS cells immediately after exposure and 18 and 36 h after the exposure cut-off was calculated by QRT-PCR. RESULTS: The cell viability of AGS cells was decreased under the exposure of 0.2 and 2 mT EMFs when compared to the control. Up-regulation of miR-144 and miR-375 were observed in AGS cells under the exposure of magnetic fields. CONCLUSIONS: The results indicated that the miR levels were significantly decreased 18 and 36 h after finishing the exposure, but not reached the normal range. The results of this investigation indicated that weak and moderate intermittent 50 Hz ELF-EMFs can induce changes in miRNA expression.


Assuntos
Campos Eletromagnéticos , MicroRNAs/genética , Neoplasias Gástricas/patologia , Regulação para Cima/efeitos da radiação , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos da radiação , Humanos , Ativação Transcricional/efeitos da radiação
13.
J Photochem Photobiol B ; 221: 112245, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34182186

RESUMO

There is currently no clear understanding on the pathways involved in the process of cell inhibition by photobiomodulation (PBM). The present study evaluated the influence of PBM on the expression of autophagy markers in vitro in an in situ model of oral carcinoma. Oral squamous cell carcinoma (Cal27) and stromal fibroblasts (FG) cultures were used. The independent variables were 'cell type' (FG and CAL27) 'culture condition' (monocultures or co-cultures) and PBM (placebo and 36 J/cm2). The cultures were irradiated from a red LED source for mRNA expression and protein expression analyses. The autophagy markers evaluated were Beclin-1, LC3B and p62 as well as adjuvant markers (BAX Bcl-2, VEGF, CD105, CD34, PRDX1, PRDX4 and GRP78). The Cal27 cells upregulated the autophagy markers upon exposure to PBM both at the mRNA and protein expression levels, providing evidence to explain malignant cell inhibition by PBM.


Assuntos
Autofagia/genética , Luz , Regulação para Cima/efeitos da radiação , Proteína Beclina-1/genética , Proteína Beclina-1/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular , Técnicas de Cocultura , Chaperona BiP do Retículo Endoplasmático , Humanos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Neoplasias Bucais/metabolismo , Neoplasias Bucais/patologia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Mensageiro/metabolismo
14.
Biomed Pharmacother ; 141: 111801, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34146850

RESUMO

Plant-based natural extracts contain several nutrients and bioactive compounds, such as phenolics and flavonoids, that possess various health-promoting activities. This study investigated the effects of polyphenols from Pterocarpus santalinus hydroalcoholic extract (PSHE) against gamma radiation-induced derangements via the upregulation of Nrf2. Ultra High Performance Liquid Chromatography Coupled to High Resolution Mass Spectrometry (UHPLC-HRMS/MS) analysis was performed to identify the possible radioprotectors. In vivo and in vitro studies, namely Real-Time-PCR (RT-PCR) analysis, Reactive Oxygen Species (ROS) scavenging activity, lipid peroxidation and GSH levels, DNA damage and cell death studies, anti-inflammatory (Sandwich ELISA), immunomodulatory studies (antibody staining), and model free radical scavenging assays, were performed. Vanillic acid, protocatechuic acid, para-hydroxybenzoic acid, chlorogenic acid, TNF-α inhibitor (Eudesmin), isoflavone (Daidzein 7-o-glucoside), astragalin (Kaempferol 3-o-glycoside), and other polyphenols were identified in PSHE using UHPLC-HRMS/MS analysis. Prophylactic administration of PSHE (-1 h) rendered more than 33% survival in mice exposed to 8 Gy whole-body-irradiation with increased mice survival and recovery of bone marrow and spleen cellularity. Real-time RT-PCR analysis showed that PSHE treatment (50 µg/mL) upregulated Nrf2, HO-1, and GPX-1 in mice splenocytes. At 50 µg/mL, PSHE reduced ROSscavenging activity, mitochondrial and spleen membrane lipid peroxidation levels, DNA damage, and cell death, and increased GSH levels. At 10 µg/mL, PSHE treatment diminished the content of IL-6 and TNF-α. At 50 µg/mL, PSHE suppressed lymphocyte proliferation. These findings indicate that polyphenols of PSHE possess marked antioxidant, anti-inflammatory, and immunomodulatory capacities, which play important roles in the prevention of radiation damage.


Assuntos
Raios gama/efeitos adversos , Fator 2 Relacionado a NF-E2/biossíntese , Fator 2 Relacionado a NF-E2/efeitos da radiação , Extratos Vegetais/farmacologia , Pterocarpus , Animais , Relação Dose-Resposta a Droga , Sequestradores de Radicais Livres/isolamento & purificação , Sequestradores de Radicais Livres/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Extratos Vegetais/isolamento & purificação , Células RAW 264.7 , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/efeitos da radiação
15.
Int J Mol Sci ; 22(9)2021 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-33946760

RESUMO

Fagopyrum tataricum 'Hokkai T10' is a buckwheat cultivar capable of producing large amounts of phenolic compounds, including flavonoids (anthocyanins), phenolic acids, and catechin, which have antioxidant, anticancer, and anti-inflammatory properties. In the present study, we revealed that the maize transcription factor Lc increased the accumulation of phenolic compounds, including sinapic acid, 4-hydroxybenzonate, t-cinnamic acid, and rutin, in Hokkai T10 hairy roots cultured under long-photoperiod (16 h light and 8 h dark) conditions. The transcription factor upregulated phenylpropanoid and flavonoid biosynthesis pathway genes, yielding total phenolic contents reaching 27.0 ± 3.30 mg g-1 dry weight, 163% greater than the total flavonoid content produced by a GUS-overexpressing line (control). In contrast, when cultured under continuous darkness, the phenolic accumulation was not significantly different between the ZmLC-overexpressing hairy roots and the control. These findings suggest that the transcription factor (ZmLC) activity may be light-responsive in the ZmLC-overexpressing hairy roots of F. tataricum, triggering activation of the phenylpropanoid and flavonoid biosynthesis pathways. Further studies are required on the optimization of light intensity in ZmLC-overexpressing hairy roots of F. tataricum to enhance the production of phenolic compounds.


Assuntos
Fagopyrum/metabolismo , Fagopyrum/efeitos da radiação , Fenóis/metabolismo , Vias Biossintéticas/genética , Vias Biossintéticas/efeitos da radiação , Escuridão , Fagopyrum/genética , Flavonoides/biossíntese , Regulação da Expressão Gênica de Plantas/efeitos da radiação , Genes de Plantas/efeitos da radiação , Luz , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Raízes de Plantas/genética , Raízes de Plantas/metabolismo , Plantas Geneticamente Modificadas , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Regulação para Cima/efeitos da radiação
16.
Sci Rep ; 11(1): 10177, 2021 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-33986387

RESUMO

As a radiation biodosimetry tool, gene expression profiling is being developed using mouse and human peripheral blood models. The impact of dose, dose-rate, and radiation quality has been studied with the goal of predicting radiological tissue injury. In this study, we determined the impact of aging on the gene expression profile of blood from mice exposed to radiation. Young (2 mo) and old (21 mo) male mice were irradiated with 4 Gy x-rays, total RNA was isolated from whole blood 24 h later, and subjected to whole genome microarray analysis. Pathway analysis of differentially expressed genes revealed young mice responded to x-ray exposure by significantly upregulating pathways involved in apoptosis and phagocytosis, a process that eliminates apoptotic cells and preserves tissue homeostasis. In contrast, the functional annotation of senescence was overrepresented among differentially expressed genes from irradiated old mice without enrichment of phagocytosis pathways. Pathways associated with hematologic malignancies were enriched in irradiated old mice compared with irradiated young mice. The fibroblast growth factor signaling pathway was underrepresented in older mice under basal conditions. Similarly, brain-related functions were underrepresented in unirradiated old mice. Thus, age-dependent gene expression differences should be considered when developing gene signatures for use in radiation biodosimetry.


Assuntos
Regulação da Expressão Gênica/genética , Exposição à Radiação , Transcriptoma/efeitos da radiação , Fatores Etários , Envelhecimento/genética , Envelhecimento/efeitos da radiação , Algoritmos , Animais , Apoptose/genética , Apoptose/efeitos da radiação , Contagem de Células Sanguíneas , Biologia Computacional , Regulação para Baixo/efeitos da radiação , Masculino , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Fagocitose/genética , Fagocitose/efeitos da radiação , Transdução de Sinais/efeitos da radiação , Regulação para Cima/efeitos da radiação , Raios X
17.
Int J Mol Med ; 47(6)2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33907823

RESUMO

Radiation is one of the main methods for the treatment of colorectal cancer (CRC) before or after surgery. However, radiotherapy tolerance of patients with CRC is often a major concern. Interferon regulatory factor 1 (IRF1) is a member of the IRF family and is involved in the development of multiple diseases, including tumors. The present study investigated the role of IRF1 in the development and radiation sensitivity of CRC. Immunohistochemistry was performed to examine the expression levels of IRF1 in tissue samples from patients with CRC, as well as in nude mice. MTT, 5­ethynyl­20­deoxyuridine, colony formation, cell cycle alteration and apoptosis assays were performed in CRC cell lines. Western blotting and immunofluorescence were used to detect the expression levels of a series of proteins. RNA sequencing was applied to identify genes whose expression was upregulated by IRF1 overexpression. Xenograft nude mouse models and hematoxylin and eosin staining were used to validate the present findings in vivo. It was revealed that the expression levels of IRF1 were significantly lower in CRC tissues than in adjacent tissues. IRF1 upregulation inhibited cell proliferation and colony formation, caused G1 cell arrest, promoted cell apoptosis, and enhanced the sensitivity of CRC cells to X­ray irradiation. The role of IRF1 in promoting the radiosensitivity of CRC was further demonstrated in nude mice with CRC xenografts. In addition, RNA sequencing revealed that overexpression of IRF1 in CRC cells significantly increased the expression levels of interferon­induced protein family members interferon α inducible protein 6, interferon induced transmembrane protein 1 and interferon induced protein 35 (fold change >2.0). In summary, the present study demonstrated that the upregulation of IRF1 inhibited the progression and promoted the radiosensitivity of CRC, likely by regulating interferon­induced proteins.


Assuntos
Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Progressão da Doença , Fator Regulador 1 de Interferon/metabolismo , Interferons/metabolismo , Proteínas de Neoplasias/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Apoptose/genética , Apoptose/efeitos da radiação , Ciclo Celular/genética , Ciclo Celular/efeitos da radiação , Linhagem Celular Tumoral , Proliferação de Células/genética , Proliferação de Células/efeitos da radiação , Neoplasias Colorretais/genética , Regulação para Baixo/genética , Regulação para Baixo/efeitos da radiação , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Tolerância a Radiação/genética , Tolerância a Radiação/efeitos da radiação , Taxa de Sobrevida , Regulação para Cima/genética , Regulação para Cima/efeitos da radiação , Raios X
18.
J Photochem Photobiol B ; 219: 112188, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33901880

RESUMO

Far-infrared (FIR) irradiation is reported to inhibit cell proliferation in various types of cancer cells; the underlying mechanism, however, remains unclear. We explored the molecular mechanisms using MDA-MB-231 human breast cancer cells. FIR irradiation significantly inhibited cell proliferation and colony formation compared to hyperthermal stimulus, with no alteration in cell viability. No increase in DNA fragmentation or phosphorylation of DNA damage kinases including ataxia-telangiectasia mutated kinase, ataxia telangiectasia and Rad3-related kinase, and DNA-dependent protein kinase indicated no DNA damage. FIR irradiation increased the phosphorylation of checkpoint kinase 2 (Chk2) at Thr68 (p-Chk2-Thr68) but not that of checkpoint kinase 1 at Ser345. Increased nuclear p-Chk2-Thr68 and Ca2+/CaM accumulations were found in FIR-irradiated cells, as observed in confocal microscopic analyses and cell fractionation assays. In silico analysis predicted that Chk2 possesses a Ca2+/calmodulin (CaM) binding motif ahead of its kinase domain. Indeed, Chk2 physically interacted with CaM in the presence of Ca2+, with their binding markedly pronounced in FIR-irradiated cells. Pre-treatment with a Ca2+ chelator significantly reversed FIR irradiation-increased p-Chk2-Thr68 expression. In addition, a CaM antagonist or small interfering RNA-mediated knockdown of the CaM gene expression significantly attenuated FIR irradiation-increased p-Chk2-Thr68 expression. Finally, pre-treatment with a potent Chk2 inhibitor significantly reversed both FIR irradiation-stimulated p-Chk2-Thr68 expression and irradiation-repressed cell proliferation. In conclusion, our results demonstrate that FIR irradiation inhibited breast cancer cell proliferation, independently of DNA damage, by activating the Ca2+/CaM/Chk2 signaling pathway in the nucleus. These results demonstrate a novel Chk2 activation mechanism that functions irrespective of DNA damage.


Assuntos
Cálcio/metabolismo , Calmodulina/metabolismo , Proliferação de Células/efeitos da radiação , Quinase do Ponto de Checagem 2/metabolismo , Dano ao DNA/efeitos da radiação , Raios Infravermelhos , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Sobrevivência Celular/efeitos da radiação , Quinase do Ponto de Checagem 2/antagonistas & inibidores , Quinase do Ponto de Checagem 2/genética , Feminino , Humanos , Fosforilação/efeitos da radiação , Ligação Proteica , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Regulação para Cima/efeitos da radiação
19.
J Photochem Photobiol B ; 216: 112147, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33561689

RESUMO

Ultraviolet B (UVB) from the sunlight is a major environmental cause for human skin damages, inducing cell death, inflammation, senescence and even carcinogenesis. The natural flavonoid silibinin, clinically used as liver protectant, has protective effects against UVB-caused skin injury in vivo and in vitro. Silibinin is often classified as a phytoestrogen, because it modulates the activation of estrogen receptors (ERs). However, whether silibinin's estrogenic effect contributes to the skin protection against UVB injury remains to be elucidated. The issue was explored in this study by using the human foreskin dermal fibroblasts (HFF) and human non-malignant immortalized keratinocytes (HaCaT). In HFF, pre-treatment with silibinin rescued UVB-irradiated cells from apoptosis. Interestingly, silibinin increased the whole cellular and nuclear levels of ERα and ERß in UVB-irradiated cells. Activation of ERs by treatment with estradiol elevated the cell survival and reduced apoptosis in UVB-treated cells. ERα agonist increased cell survival, while its antagonist decreased it. ERß agonist also increased cell survival, but the antagonist had no effect on cell survival. Transfection of the cells with the small interfering RNAs (si-RNAs) to ERα or ERß diminished the protective effect of silibinin on UVB-irradiated cells. In UVB-treated HaCaT cells, both ERα and ERß were increased by silibinin treatment. Inhibition of activation and expression of ERα or ERß by specific antagonists and si-RNAs, respectively, reduced cell survival in UVB-treated HaCaT cells regardless of silibinin treatment. Taken together, it is summarized that silibinin up-regulates both ERα and ERß pathways in UVB-treated dermal HFF cells and epidermal HaCaT cells, leading to protection of skin from UVB-damage.


Assuntos
Fibroblastos/efeitos da radiação , Substâncias Protetoras/química , Receptores de Estrogênio/antagonistas & inibidores , Silibina/química , Apoptose/efeitos da radiação , Fibroblastos/citologia , Células HaCaT , Humanos , Substâncias Protetoras/farmacologia , RNA Interferente Pequeno/metabolismo , RNA Interferente Pequeno/efeitos da radiação , Receptores de Estrogênio/efeitos da radiação , Transdução de Sinais , Silibina/farmacologia , Pele , Raios Ultravioleta , Regulação para Cima/efeitos da radiação
20.
Cells ; 10(1)2021 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-33445526

RESUMO

The current treatment strategy for patients with aggressive colorectal cancer has been hampered by resistance to radiotherapy and chemotherapy due to the existence of cancer stem-like cells (CSCs). Recent studies have shown that SOX2 expression plays an important role in the maintenance of CSC properties in colorectal cancer. In this study, we investigated the induction and regulatory role of SOX2 following the irradiation of radioresistant and radiosensitive colorectal cancer cells. We used FACS and western blotting to analyze SOX2 expression in cells. Among the markers of colorectal CSCs, the expression of CD44 increased upon irradiation in radioresistant cells. Further analysis revealed the retention of CSC properties with an upregulation of SOX2 as shown by enhanced resistance to radiation and metastatic potential in vitro. Interestingly, both the knockdown and overexpression of SOX2 led to increase in CD44+ population and induction of CSC properties in colorectal cancer following irradiation. Furthermore, selective genetic and pharmacological inhibition of the PI3K/AKT pathway, but not the MAPK pathway, attenuated SOX2-dependent CD44 expression and metastatic potential upon irradiation in vitro. Our findings suggested that SOX2 regulated by radiation-induced activation of PI3K/AKT pathway contributes to the induction of colorectal CSCs, thereby highlighting its potential as a therapeutic target.


Assuntos
Células-Tronco Neoplásicas/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Radiação , Fatores de Transcrição SOXB1/metabolismo , Transdução de Sinais , Regulação para Cima , Linhagem Celular Tumoral , Movimento Celular/efeitos da radiação , Humanos , Receptores de Hialuronatos/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos da radiação , Invasividade Neoplásica , Células-Tronco Neoplásicas/efeitos da radiação , Tolerância a Radiação/efeitos da radiação , Transdução de Sinais/efeitos da radiação , Regulação para Cima/efeitos da radiação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA